I Discovery and Modulation of Acid-Sensing Ion

Total Page:16

File Type:pdf, Size:1020Kb

I Discovery and Modulation of Acid-Sensing Ion Discovery and modulation of acid-sensing ion channel modulating venom peptides Ben Cristofori-Armstrong Bachelor of Science (Hons) A thesis submitted for the degree of Doctor of Philosophy at The University of Queensland in 2018 Faculty of Medicine, School of Biomedical Sciences I Abstract Acid-sensing ion channels (ASICs) are a family of proton-activated cation channels expressed in a variety of neuronal and non-neuronal tissues. As proton-gated channels, they have been implicated in many pathophysiological conditions where pH is perturbed. Venom derived compounds represent the most potent and selective modulators of ASICs described to date, and thus have been invaluable pharmacological tools to study ASIC structure, function, and biological roles. There are now eleven ASIC modulators described from animal venoms, with those from snakes and spiders favouring ASIC1, while the sea anemone and cone snail modulators preferentially target ASIC3. Chapter 1 reviews the current state of knowledge on venom derived ASIC modulators, with a particular focus on their molecular interaction with ASICs, what they have taught us about channel structure, and what they may still reveal about ASIC function and pathophysiological roles. Venom peptides are often disulfide bonded making their recombinant expression challenging. Chapter 2 describes a periplasmic Escherichia coli protocol for production of correctly folded peptides that was used throughout this thesis. Using the two-electrode voltage-clamp technique, Chapter 3 shows that a wide variety of voltage- and ligand-gated ion channels have the same channel properties and pharmacological profiles when expressed in either Xenopus laevis or X. borealis oocytes. This voltage-clamp technique was heavily used throughout this thesis to perform functional studies of venom peptides. The spider venom peptide PcTx1 is the best studied ASIC modulator; it has an IC50 of ~1 nM at rat ASIC1a and is neuroprotective in rodent models of ischemic stroke. Chapter 4 examines the molecular interaction between PcTx1 and both human ASIC1a and the off- target ASIC1b subtype, where little experimental work has been done. We show that although PcTx1 is 10-fold less potent at human ASIC1a than the rat channel, the apparent affinity for the two channels is comparable. The pharmacophore of PcTx1 for human ASIC1a and rat ASIC1b was examined via alanine scanning mutagenesis and uncovered residues that show subtle ASIC1 species and subtype-dependent differences in activity that may allow for further manipulation to develop more selective PcTx1 analogues. The ASIC3 inhibitor APETx2 is analgesic in rodent models of chemically-induced, inflammatory and postoperative pain, providing strong evidence for the ASIC3 subtype as a potential pain target. Despite the comprehensive structure-activity studies of APETx2, the mechanism of action and channel binding site have remained elusive. Chapter 5 fills this II gap in knowledge of an important ASIC research tool and also reveals subtle differences in the pharmacophore of APETx2 for inhibition of ASIC3 and potentiation of ASIC1b. We propose that APETx2 binds to a novel region for peptide modulators of ASICs and acts by preventing the closed-to-open gating transition of rat ASIC3. Mambalgins are a group of three-finger toxins isolated from black and green mamba snake venoms that target ASIC1a and ASIC1b containing channels. Unique among ASIC modulators, the potent inhibitory activity of mambalgins at rodent ASIC1b was crucial in demonstrating that ASIC1b, and not ASIC1a, is important for peripheral pain sensing in rodents. Chapter 6 shows that the efficacy of mambalgins varies between the ASIC1 splice variants ASIC1a and ASIC1b, in both human and rat channels. Strikingly, mambalgin is a potentiator of human ASIC1b under certain conditions. Furthermore, these pharmacological differences are due to both the molecular interactions at the binding sites and the different ways mambalgin can modify the gating characteristics of specific ASIC variants. Chapters 7 describes the isolation, pharmacological characterisation and chemical stability of the novel spider venom peptide Hm3a from the venom of the tarantula Heteroscodra maculata. Hm3a is a close homolog of PcTx1 with five amino acid substitutions and a three residue C-terminal truncation. Despite its high sequence similarity with PcTx1 and similar pharmacology, Hm3a showed higher levels of stability over 48 h and will be particularly useful when stability in biological fluids is required, for example in long term in vitro cell- based assays and in vivo experiments. Chapter 8 describes the characterisation of conorfamides As1a and As2a from the venom of the cone snail Conus austini. Amidated peptide variants altered desensitization of ASIC1a and 3, and a lysine to arginine mutation introduced ASIC1a peak current potentiation. These conorfamides also inhibited a7 and muscle-type nicotinic acetylcholine receptors (nAChR) at nanomolar concentrations. These are the first conorfamides with the dual pharmacology described to date. III Declaration by author This thesis is composed of my original work, and contains no material previously published or written by another person except where due reference has been made in the text. I have clearly stated the contribution by others to jointly-authored works that I have included in my thesis. I have clearly stated the contribution of others to my thesis as a whole, including statistical assistance, survey design, data analysis, significant technical procedures, professional editorial advice, financial support and any other original research work used or reported in my thesis. The content of my thesis is the result of work I have carried out since the commencement of my higher degree by research candidature and does not include a substantial part of work that has been submitted to qualify for the award of any other degree or diploma in any university or other tertiary institution. I have clearly stated which parts of my thesis, if any, have been submitted to qualify for another award. I acknowledge that an electronic copy of my thesis must be lodged with the University Library and, subject to the policy and procedures of The University of Queensland, the thesis be made available for research and study in accordance with the Copyright Act 1968 unless a period of embargo has been approved by the Dean of the Graduate School. I acknowledge that copyright of all material contained in my thesis resides with the copyright holder(s) of that material. Where appropriate I have obtained copyright permission from the copyright holder to reproduce material in this thesis and have sought permission from co- authors for any jointly authored works included in the thesis. IV Publications included in this thesis 1. B Cristofori-Armstrong# and LD Rash# (2017) Acid-sensing ion channel (ASIC) structure and function: Insights from spider, snake and sea anemone. Neuropharmacology 127: 173–184. 2. SY Er*, B Cristofori-Armstrong*, P Escoubas and LD Rash (2017) Discovery and molecular interaction studies of a highly stable, tarantula peptide modulator of acid- sensing ion channel 1. Neuropharmacology 127: 185–195. 3. B Cristofori-Armstrong, MS Soh, S Talwar, DL Brown, JDO Griffin, Z Dekan, JL Stow, GF King, JW Lynch and LD Rash (2015) Xenopus borealis as an alternative source of oocytes for biophysical and pharmacological studies of neuronal ion channels. Scientific Reports 5: 14763. V Submitted manuscripts included in this thesis 1. B Cristofori-Armstrong*, NJ Saez*, IR Chassagnon, GF King, and LD Rash. The modulation of acid-sensing ion channel 1 by PcTx1 is pH-, subtype- and species- dependent: importance of interactions at the channel subunit interface and potential for engineering selective analogues. Biochemical Pharmacology. Revisions resubmitted. 2. A-H Jin*, B Cristofori-Armstrong*, LD Rash, SAR González, RJ Lewis, PF Alewood, and I Vetter. Novel conorfamides from Conus austini venom modulate both nicotinic acetylecholine receptors and acid-sensing ion channels. Biochemical Pharmacology. Submitted. Other publications during candidature Book Chapter 1. NJ Saez, B Cristofori-Armstrong, R Anangi, and GF King (2017) A Strategy for Production of Correctly Folded Disulfide-rich Peptides in the Periplasm of E. coli. Methods in Molecular Biology 1586: 155–180. Journal Article (* joint first author; # co-corresponding author) 1. A Silva*, B Cristofori-Armstrong*, LD Rash, WC Hodgson and GK Isbister (2018) Defining the role of post-synaptic -neurotoxins in paralysis due to envenoming in humans. Cellular and Molecular Life Sciences 75: 4465–4478 2. H Shen, Z Li, Y Jiang, X Pan, J Wu, B Cristofori-Armstrong, JJ Smith, YKY Chin, J Lei, Q Zhou, GF King, N Yan (2018) Structural basis for the modulation of voltage-gated sodium channels by animal toxins. Science 362: eaau2596 3. B Madio, S Peigneur, YKY Chin, BR Hamilton, ST Henriques, JJ Smith, B Cristofori- Armstrong, Z Dekan, BA Boughton, PF Alewood, J Tytgat, GF King and EAB Undheim (2018) PHAB toxins: a unique family of sea anemone toxins evolving via intra-gene concerted evolution defines a new peptide fold. Cellular and Molecular Life Science 75: 4511–4524. 4. JYP Lee*, NJ Saez*, B Cristofori-Armstrong*, R Anangi, GF King, MT Smith and LD Rash (2018) Inhibition of acid-sensing ion channels by diminaene and APETx2 evoke VI partial and highly variable antihyperalgesia in a rat model of inflammatory pain. British Journal of Pharmacology 175: 2204–2218. 5. SS Pineda, PA Chaumeil, A Kunert, Q Kaas, MWC Thang, L Li, M Nuhm, V Herzig, NJ Saez, B Cristofori-Armstrong, R Anangi, S Senff, D Gorse and GF King (2017) ArachnoServer 3.0: an online resource for automated discovery, analysis and annotation of spider toxins. Bioinformatics 34: 1074–1076. 6. JS Wingerd, CA Mozar, CA Ussing, SS Murali, YKY Chin, B Cristofori-Armstrong, T Durek, J Gilchrist, CW Vaughan, F Bosman, DJ Adams, RJ Lewis, PF Alewood, M Mobli, MJ Christie and LD Rash (2017) The tarantula toxin /-TRTX-Pre1a highlights the importance of the S1-S2 voltage-sensor region for sodium channel subtype selectivity. Scientific Reports 7: 974.
Recommended publications
  • Biological Toxins Fact Sheet
    Work with FACT SHEET Biological Toxins The University of Utah Institutional Biosafety Committee (IBC) reviews registrations for work with, possession of, use of, and transfer of acute biological toxins (mammalian LD50 <100 µg/kg body weight) or toxins that fall under the Federal Select Agent Guidelines, as well as the organisms, both natural and recombinant, which produce these toxins Toxins Requiring IBC Registration Laboratory Practices Guidelines for working with biological toxins can be found The following toxins require registration with the IBC. The list in Appendix I of the Biosafety in Microbiological and is not comprehensive. Any toxin with an LD50 greater than 100 µg/kg body weight, or on the select agent list requires Biomedical Laboratories registration. Principal investigators should confirm whether or (http://www.cdc.gov/biosafety/publications/bmbl5/i not the toxins they propose to work with require IBC ndex.htm). These are summarized below. registration by contacting the OEHS Biosafety Officer at [email protected] or 801-581-6590. Routine operations with dilute toxin solutions are Abrin conducted using Biosafety Level 2 (BSL2) practices and Aflatoxin these must be detailed in the IBC protocol and will be Bacillus anthracis edema factor verified during the inspection by OEHS staff prior to IBC Bacillus anthracis lethal toxin Botulinum neurotoxins approval. BSL2 Inspection checklists can be found here Brevetoxin (http://oehs.utah.edu/research-safety/biosafety/ Cholera toxin biosafety-laboratory-audits). All personnel working with Clostridium difficile toxin biological toxins or accessing a toxin laboratory must be Clostridium perfringens toxins Conotoxins trained in the theory and practice of the toxins to be used, Dendrotoxin (DTX) with special emphasis on the nature of the hazards Diacetoxyscirpenol (DAS) associated with laboratory operations and should be Diphtheria toxin familiar with the signs and symptoms of toxin exposure.
    [Show full text]
  • Animal Venom Derived Toxins Are Novel Analgesics for Treatment Of
    Short Communication iMedPub Journals 2018 www.imedpub.com Journal of Molecular Sciences Vol.2 No.1:6 Animal Venom Derived Toxins are Novel Upadhyay RK* Analgesics for Treatment of Arthritis Department of Zoology, DDU Gorakhpur University, Gorakhpur, UP, India Abstract *Corresponding authors: Ravi Kant Upadhyay Present review article explains use of animal venom derived toxins as analgesics of the treatment of chronic pain and inflammation occurs in arthritis. It is a [email protected] progressive degenerative joint disease that put major impact on joint function and quality of life. Patients face prolonged inappropriate inflammatory responses and bone erosion. Longer persistent chronic pain is a complex and debilitating Department of Zoology, DDU Gorakhpur condition associated with a large personal, mental, physical and socioeconomic University, Gorakhpur, UttarPradesh, India. burden. However, for mitigation of inflammation and sever pain in joints synthetic analgesics are used to provide quick relief from pain but they impose many long Tel: 9838448495 term side effects. Venom toxins showed high affinity to voltage gated channels, and pain receptors. These are strong inhibitors of ion channels which enable them as potential therapeutic agents for the treatment of pain. Present article Citation: Upadhyay RK (2018) Animal Venom emphasizes development of a new class of analgesic agents in form of venom Derived Toxins are Novel Analgesics for derived toxins for the treatment of arthritis. Treatment of Arthritis. J Mol Sci. Vol.2 No.1:6 Keywords: Analgesics; Venom toxins; Ion channels; Channel inhibitors; Pain; Inflammation Received: February 04, 2018; Accepted: March 12, 2018; Published: March 19, 2018 Introduction such as the back, spine, and pelvis.
    [Show full text]
  • Slow Inactivation in Voltage Gated Potassium Channels Is Insensitive to the Binding of Pore Occluding Peptide Toxins
    Biophysical Journal Volume 89 August 2005 1009–1019 1009 Slow Inactivation in Voltage Gated Potassium Channels Is Insensitive to the Binding of Pore Occluding Peptide Toxins Carolina Oliva, Vivian Gonza´lez, and David Naranjo Centro de Neurociencias de Valparaı´so, Facultad de Ciencias, Universidad de Valparaı´so, Valparaı´so, Chile ABSTRACT Voltage gated potassium channels open and inactivate in response to changes of the voltage across the membrane. After removal of the fast N-type inactivation, voltage gated Shaker K-channels (Shaker-IR) are still able to inactivate through a poorly understood closure of the ion conduction pore. This, usually slower, inactivation shares with binding of pore occluding peptide toxin two important features: i), both are sensitive to the occupancy of the pore by permeant ions or tetraethylammonium, and ii), both are critically affected by point mutations in the external vestibule. Thus, mutual interference between these two processes is expected. To explore the extent of the conformational change involved in Shaker slow inactivation, we estimated the energetic impact of such interference. We used kÿconotoxin-PVIIA (kÿPVIIA) and charybdotoxin (CTX) peptides that occlude the pore of Shaker K-channels with a simple 1:1 stoichiometry and with kinetics 100-fold faster than that of slow inactivation. Because inactivation appears functionally different between outside-out patches and whole oocytes, we also compared the toxin effect on inactivation with these two techniques. Surprisingly, the rate of macroscopic inactivation and the rate of recovery, regardless of the technique used, were toxin insensitive. We also found that the fraction of inactivated channels at equilibrium remained unchanged at saturating kÿPVIIA.
    [Show full text]
  • Α-Dendrotoxin Inhibits the ASIC Current in Dorsal Root Ganglion
    Neuroscience Letters 606 (2015) 42–47 Contents lists available at ScienceDirect Neuroscience Letters journal homepage: www.elsevier.com/locate/neulet Research paper ␣-Dendrotoxin inhibits the ASIC current in dorsal root ganglion neurons from rat a a b c b Adriana Báez , Emilio Salceda , Martín Fló , Martín Grana˜ , Cecilia Fernández , a a,∗ Rosario Vega , Enrique Soto a Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), 14 sur 6301, CU, San Manuel, Puebla, Pue., CP 72570, Mexico b Faculty of Chemistry, UDELAR, Av. Gral., Flores, Montevideo 2124, Uruguay c Institut Pasteur de Montevideo, Mataojo 2020, Montevideo 11400, Uruguay h i g h l i g h t s g r a p h i c a l a b s t r a c t • ␣-DTx inhibits the peak amplitude of proton gated current (ASIC) in DRG neurons. • ␣-DTx action on ASICs is reversible and dose dependent. • ␣-DTx inhibits ASIC currents with IC50 in the nM range. • ␣-DTx positive charge at neutral pH is similar to other components inhibit- ing ASICs. • Other toxins considered to be highly selective may have dual action such as ␣-DTx. a r t i c l e i n f o a b s t r a c t Article history: Dendrotoxins are a group of peptide toxins purified from the venom of several mamba snakes. ␣- Received 27 April 2015 Dendrotoxin (␣-DTx, from the Eastern green mamba Dendroaspis angusticeps) is a well-known blocker Received in revised form 11 August 2015 + of voltage-gated K channels and specifically of Kv1.1, Kv1.2 and Kv1.6.
    [Show full text]
  • Towards Therapeutic Applications of Arthropod Venom K+-Channel Blockers in CNS Neurologic Diseases Involving Memory Acquisition and Storage
    Hindawi Publishing Corporation Journal of Toxicology Volume 2012, Article ID 756358, 21 pages doi:10.1155/2012/756358 Review Article Towards Therapeutic Applications of Arthropod Venom K+-Channel Blockers in CNS Neurologic Diseases Involving Memory Acquisition and Storage Christiano D. C. Gati,1, 2 Marcia´ R. Mortari,1 and Elisabeth F. Schwartz1 1 Departamento de Ciˆencias Fisiologicas,´ Instituto de Ciˆencias Biologicas,´ Universidade de Bras´ılia, 70910-900 Bras´ılia, DF, Brazil 2 Universidade Catolica´ de Bras´ılia, 71966-700 Bras´ılia, DF, Brazil Correspondence should be addressed to Elisabeth F. Schwartz, [email protected] Received 29 December 2011; Accepted 8 February 2012 Academic Editor: Yonghua Ji Copyright © 2012 Christiano D. C. Gati et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Potassium channels are the most heterogeneous and widely distributed group of ion channels and play important functions in all cells, in both normal and pathological mechanisms, including learning and memory processes. Being fundamental for many diverse physiological processes, K+-channels are recognized as potential therapeutic targets in the treatment of several Central Nervous System (CNS) diseases, such as multiple sclerosis, Parkinson’s and Alzheimer’s diseases, schizophrenia, HIV-1-associated dementia, and epilepsy. Blockers of these channels are therefore potential candidates for the symptomatic treatment of these neuropathies, through their neurological effects. Venomous animals have evolved a wide set of toxins for prey capture and defense. These compounds, mainly peptides, act on various pharmacological targets, making them an innumerable source of ligands for answering experimental paradigms, as well as for therapeutic application.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2014/0088056A1 Ye Et Al
    US 20140O88056A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2014/0088056A1 Ye et al. (43) Pub. Date: Mar. 27, 2014 (54) CARDIAC GLYCOSIDES ARE POTENT Publication Classification INHIBITORS OF INTERFERON-BETA GENE EXPRESSION (51) Int. Cl. A613 L/585 (2006.01) A 6LX3/59 (2006.01) (75) Inventors: Junqiang Ye, Fort Lee, NJ (US); A613 L/7 (2006.01) Shuibing Chen, Pelham, NY (US); Tom A 6LX3 L/505 (2006.01) Maniatis, New York, NY (US) A613 L/407 (2006.01) A63L/36 (2006.01) (52) U.S. Cl. (73) Assignee: PRESIDENT AND FELLOWS OF CPC ............. A6 IK3I/585 (2013.01); A61 K3I/407 HARVARD COLLEGE, Cambridge, (2013.01); A61 K3I/136 (2013.01); A61 K MA (US) 3 1/17 (2013.01); A61K3I/505 (2013.01); A6 IK3I/519 (2013.01) (21) Appl. No.: 13/876,795 USPC ........... 514/175: 514/410; 514/656; 514/597; (22) PCT Filed: Sep. 28, 2011 514/256; 514/264.11: 435/375; 435/184 (57) ABSTRACT (86). PCT No.: The invention provides for a method of inhibiting interferon S371 (c)(1), beta gene expression and/or reducing the level of interferon (2), (4) Date: Nov. 25, 2013 beta in a cell by contacting the cell with a Na", Ca", or K" ion-channel modulator. The invention also provides for a method of treating a disease or disorder characterized by Related U.S. Application Data elevated interferon beta levels or elevated levels of interferon (60) Provisional application No. 61/387,407, filed on Sep. beta gene expression. Additionally, the invention provides a 28, 2010.
    [Show full text]
  • (12) United States Patent (10) Patent No.: US 9.248,185 B2 Rubin Et Al
    USOO9248185B2 (12) United States Patent (10) Patent No.: US 9.248,185 B2 Rubin et al. (45) Date of Patent: Feb. 2, 2016 (54) METHODS OF INCREASING SATELLITE (2013.01); A61 K3I/485 (2013.01); A61 K CELL PROLIFERATION 3 I/553 (2013.01); A61 K3I/58 (2013.01); A61K3I/7076 (2013.01); G0IN33/5044 (75) Inventors: Lee L. Rubin, Wellesley, MA (US); (2013.01); C07D498/22 (2013.01) Amanda Gee, Alexandria, VA (US); (58) Field of Classification Search Amy J. Wagers, Cambridge, MA (US) None (73) Assignee: President and Fellows of Harvard See application file for complete search history. College, Cambridge, MA (US) (56) References Cited (*) Notice: Subject to any disclaimer, the term of this U.S. PATENT DOCUMENTS patent is extended or adjusted under 35 U.S.C. 154(b) by 0 days. 2003/018151.0 A1* 9, 2003 Baker et al. ................... 514,432 2005/0281788 A1 12/2005 de Bari et al. (21) Appl. No.: 14/126,716 2010.0048534 A1 2/2010 Dziki et al. .............. 514, 21108 (22) PCT Filed: Jun. 18, 2012 FOREIGN PATENT DOCUMENTS (86). PCT No.: PCT/US2O12/042964 RU 2368398 9, 2009 S371 (c)(1), OTHER PUBLICATIONS (2), (4) Date: Jun. 13, 2014 Shea et al (2010. Cell Stem Cell. 6: 117-129).* Strocket al., 2003. Cancer Research. 63:5559-5563.* (87) PCT Pub. No.: WO2012/174537 Mulligan et al., 2004. Nature Reviews: Cancer, 14: 173-186.* PCT Pub. Date: Dec. 20, 2012 Carlson, et al. “Relative roles of TGF-B1 and Wnt in the systemic regulation and aging of Satellite cell responses'.
    [Show full text]
  • Item Name Catalog Number Quantity A-803467 A-105 10 Mg A
    Catalog Item Name Number Quantity A-803467 A-105 10 mg A-803467 A-105 25 mg A-803467 A-105 50 mg A-803467 A-105 100 mg Amlodipine A-110 250 mg Amlodipine A-110 1 g Amlodipine A-110 5 g Amlodipine A-110 10 g 4-Aminopyridine A-115 25 g 4-Aminopyridine A-115 100 g Amlodipine besylate A-120 10 mg Amlodipine besylate A-120 25 mg Amlodipine besylate A-120 50 mg Amlodipine besylate A-120 100 mg Amlodipine besylate A-120 500 mg Amlodipine besylate A-120 1 g Amlodipine besylate A-120 5 g Azelnidipine A-135 5 mg Azelnidipine A-135 10 mg Azelnidipine A-135 25 mg Azelnidipine A-135 50 mg Amiloride hydrochloride A-140 1 g Amiloride hydrochloride A-140 5 g Amiloride hydrochloride A-140 10 g Ambroxol hydrochloride A-145 1 g Ambroxol hydrochloride A-145 5 g Aconitine A-150 25 mg Aconitine A-150 50 mg Aconitine A-150 100 mg Aconitine A-150 250 mg Amitriptyline hydrochloride A-155 10 g Amitriptyline hydrochloride A-155 25 g Amitriptyline hydrochloride A-155 100 g Amentoflavone A-165 1 mg Amentoflavone A-165 5 mg Amentoflavone A-165 10 mg Amentoflavone A-165 25 mg AMG 9810 A-180 5 mg AMG 9810 A-180 10 mg AMG 9810 A-180 25 mg AMG 9810 A-180 50 mg AM 404 A-190 5 mg AM 404 A-190 10 mg AM 404 A-190 25 mg AM 404 A-190 50 mg A-889425 A-195 1 mg A-889425 A-195 5 mg A-889425 A-195 10 mg A-889425 A-195 25 mg A-889425 A-195 50 mg 3-AQC A-205 5 mg 3-AQC A-205 10 mg 3-AQC A-205 25 mg 3-AQC A-205 50 mg ANA-12 A-215 5 mg ANA-12 A-215 10 mg ANA-12 A-215 25 mg ANA-12 A-215 50 mg ANA-12 A-215 100 mg A 967079 A-225 5 mg A 967079 A-225 10 mg A 967079 A-225 25 mg A 967079
    [Show full text]
  • Marine Toxins Targeting Kv1 Channels: Pharmacological Tools and Therapeutic Scaffolds
    marine drugs Review Marine Toxins Targeting Kv1 Channels: Pharmacological Tools and Therapeutic Scaffolds 1,2, 3, 3, Rocio K. Finol-Urdaneta * , Aleksandra Belovanovic y, Milica Micic-Vicovac y, Gemma K. Kinsella 4, Jeffrey R. McArthur 1 and Ahmed Al-Sabi 3,* 1 Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia; jeff[email protected] 2 Electrophysiology Facility for Cell Phenotyping and Drug Discovery, Wollongong, NSW 2522, Australia 3 College of Engineering and Technology, American University of the Middle East, Kuwait; [email protected] (A.B.); [email protected] (M.M.-V.) 4 School of Food Science and Environmental Health, College of Sciences and Health, Technological University Dublin, D07 ADY7 Dublin, Ireland; [email protected] * Correspondence: rfi[email protected] (R.K.F.-U.); [email protected] (A.A.-S.); Tel.: +965-22251400 (ext.1798) (A.A.-S.) These authors contributed equally to this review. y Received: 3 February 2020; Accepted: 16 March 2020; Published: 20 March 2020 Abstract: Toxins from marine animals provide molecular tools for the study of many ion channels, including mammalian voltage-gated potassium channels of the Kv1 family. Selectivity profiling and molecular investigation of these toxins have contributed to the development of novel drug leads with therapeutic potential for the treatment of ion channel-related diseases or channelopathies. Here, we review specific peptide and small-molecule marine toxins modulating Kv1 channels and thus cover recent findings of bioactives found in the venoms of marine Gastropod (cone snails), Cnidarian (sea anemones), and small compounds from cyanobacteria.
    [Show full text]
  • Electrophysiological, Pharmacological and Molecular Profile of the Transient Outward Rectifying Conductance in Rat Sympathetic Preganglionic Neurons in Vitro
    Neuroscience 178 (2011) 68–81 ELECTROPHYSIOLOGICAL, PHARMACOLOGICAL AND MOLECULAR PROFILE OF THE TRANSIENT OUTWARD RECTIFYING CONDUCTANCE IN RAT SYMPATHETIC PREGANGLIONIC NEURONS IN VITRO A. D. WHYMENT,a,b E. CODERRE,c J. M. M. WILSON,c Key words: A-current, KChIP, DPP6, rtPCR, patch clamp, L. P. RENAUD,c E. O’HAREd AND D. SPANSWICKa,b* spinal cord. aWarwick Medical School, University of Warwick, Coventry, CV4 7AL, UK bNeuroSolutions Ltd., Coventry, CV4 7ZS, UK Outward currents through potassium (Kϩ) channels are cNeurosciences Program, Ottawa Hospital Research Institute and Depart- the primary means by which excitable cells oppose mem- ment of Medicine, University of Ottawa, Ottawa, Ontario, K1Y 4E9, Canada brane excitability. Several distinct types of Kϩ-mediated dSchool of Psychology, Queen’s University Belfast, Belfast, Northern conductances have been identified in sympathetic pregan- Ireland, UK glionic neurons (SPN; Polosa et al., 1988; Pickering et al., 1991; Sah and McLachlan, 1995; Miyazaki et al., 1996; Wilson et al., 2002). One such conductance, the transient Abstract—Transient outward rectifying conductances or A-like conductances in sympathetic preganglionic neurons (SPN) are outward rectifying conductance or current (ITR), is a hall- prolonged, lasting for hundreds of milliseconds to seconds and mark of SPN and is used as an identifiable, characteristic are thought to play a key role in the regulation of SPN firing electrophysiological feature of these neurons (Yoshimura frequency. Here, a multidisciplinary electrophysiological, pharma- et al., 1987; Pickering et al., 1991; Inokuchi et al., 1993). cological and molecular single-cell rt-PCR approach was used to This conductance has historically been referred to as the ؉ investigate the kinetics, pharmacological profile and putative K A-like conductance in SPN, based upon its similarity to channel subunits underlying the transient outward rectifying con- A-type Kϩ currents and conductances described in other ductance expressed in SPN.
    [Show full text]
  • Effects of the Potassium Channel Blocking Dendrotoxins On
    Br. J. Pharmacol. (1988), 93, 215-221 Effects ofthe potassium channel blocking dendrotoxins on acetylcholine release and motor nerve terminal activity 'A.J. Anderson & A.L. Harvey Department of Physiology and Pharmacology, University of Strathclyde, Glasgow GI I XW 1 The effects of the K+ channel blocking toxins, the dendrotoxins, on neuromuscular transmission and motor nerve terminal activity were assessed on frog cutaneous pectoris, mouse diaphragm and mouse triangularis sterni nerve-muscle preparations. Endplate potentials (e.p.ps) and miniature e.p.ps were recorded with intracellular microelectrodes, and nerve terminal spikes were recorded with extracellular electrodes placed in the perineural sheaths of motor nerves. 2 Dendrotoxin from green mamba (Dendroaspis angusticeps) venom and toxin I from black mamba (D. polylepis) venom increased the amplitude of e.p.ps by increasing quantal content, and also induced repetitive e.p.ps. 3 Perineural recordings revealed that dendrotoxins could decrease the component of the waveform associated with K+ currents at the nerve terminals, and induce repetitive activation of nerve terminals. 4 In frog motor nerves, dendrotoxins are known to block the fast f, component of the K+ current at nodes of Ranvier. Blockade of a similar component ofthe K+ current at motor nerve terminals may be responsible for the effects of these toxins on neuromuscular transmission. 5 Similar conclusions can be drawn from the results obtained from mouse neuromuscular junctions. Introduction Neuronal membranes contain several distinct K+ current activated between -80 mV and -30 mV; f2, a conductances that can be distinguished by their vol- fast current activated between -40 mV and + 30 mV; tage-dependency, kinetic properties, Ca2" sensitivity, and a slow current (see Dubois, 1983).
    [Show full text]
  • Central Action of Dendrotoxin: Selective Reduction of a Transient K
    Proc. Nati. Acad. Sci. USA Vol. 83, pp. 493-497, January 1986 Neurobiology Central action of dendrotoxin: Selective reduction of a transient K conductance in hippocampus and binding to localized acceptors (K channels/facilitatory neurotoxin/neuronal binding protein) JAMES V. HALLIWELL*, IEKHSAN B. OTHMANt, ANNEGRET PELCHEN-MATTHEWSt, AND J. OLIVER DOLLYtt *Medical Research Council Neuropharmacology Research Group, School of Pharmacy, London WC1 lAX, and tDepartment of Biochemistry, Imperial College, London SW7 2AZ, United Kingdom Communicated by Sir Andrew Huxley, September 3, 1985 ABSTRACT Dendrotoxin, a small single-chain protein MATERIALS AND METHODS from the venom of Dendroaspis angusticeps, is highly toxic Purification and Radioiodination of DTX. The neurotoxin following intracerebroventricular injection into rats. Voltage- was purified to homogeneity from the venom of D. angusti- clamp analysis of CA1 neurons in hippocampal slices, treated ceps (Sigma), characterized as detailed (8) and 'l25-iodinated with tetrodotoxin, revealed that nanomolar concentrations of using a modification of the chloramine-T method that avoids dendrotoxin reduce selectively a transient, voltage-dependent loss of neurotoxicity (2). K conductance. Epileptiform activity known to be induced by Electrophysiological Recording in Hippocampal Slices. dendrotoxin can be attributed to such an action. Membrane Methods for preparation and maintenance ofbrain slices from currents not affected directly by the toxin include (i) Ca- guinea pig or rat have been described (9, 10). DTX and activated K conductance; (it) noninactivating voltage-depen- 4-aminopyridine (4AP) were dissolved in Krebs medium and dent K conductance; (Wi) inactivating and noninactivating Ca administered for required periods by superfusion at 280C. conductances; (iv) persistent inward (anomalous) rectifier Cells in the CA1 region of the slice were impaled with glass current.
    [Show full text]